effects of a bioactive scaffold containing a …...ii! effects of a bioactive scaffold containing a...

57
Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation of Stem Cells from the Apical Papilla by Craig D. Bellamy A thesis submitted in conformity with the requirements for the degree of Master of Science Faculty of Dentistry, School of Graduate Studies University of Toronto © Copyright by Craig D. Bellamy 2016

Upload: others

Post on 06-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation of Stem Cells from the Apical Papilla

by

Craig D. Bellamy

A thesis submitted in conformity with the requirements for the degree of Master of Science

Faculty of Dentistry, School of Graduate Studies University of Toronto

© Copyright by Craig D. Bellamy 2016

Page 2: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

 

ii  

Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-

Releasing Nanoparticle System on the Migration and

Differentiation of Stem Cells from the Apical Papilla

Craig D. Bellamy

Master of Science

Faculty of Dentistry, School of Graduate Studies University of Toronto

2016

Abstract The study aimed to develop and characterize a novel chitosan-based scaffold (CMCS) containing

TGF-β1-releasing chitosan nanoparticles (TGF-β1-CSnp) to enhance migration and

differentiation of SCAP. Part I concerned synthesis and characterization of the scaffold (CMCS)

and TGF-β1-CSnp. Part II examined the effect of sustained TGF-β1 release from scaffold

containing TGF-β1-CSnp on odontogenic differentiation of SCAP. The scaffold demonstrated

properties conducive to cellular activities. Incorporation of TGF-β1 in CSnp allowed sustained

release of TGF-β1 facilitating delivery of a critical concentration of TGF-β1 at the opportune

time. SCAP showed greater viability, migration and biomineralization in the presence of TGF-

β1-CSnp than in the presence of Free TGF-β1. SCAP cultured in TGF-β1-CSnp + scaffold

showed significantly higher dentin matrix protein (DMP)-1 and dentin sialophosphoprotein

(DSPP) signals compared to Free TGF-β1 + scaffold or CSnp + scaffold. These experiments

highlighted the potential of a CMCS based scaffold with growth factor releasing nanoparticles to

promote migration and differentiation of SCAP.  

Page 3: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

 

iii  

Acknowledgments

The author gratefully acknowledges the contributions of the following individuals:

Dr. Anil Kishen, primary supervisor, for his intelligence, scientific expertise, tireless

encouragement, and constant availability throughout all stages of my research, study execution,

thesis development and endodontic education.

Dr. Calvin Torneck, for sharing his knowledge and unique perspective, which had direct impact

on the study design and final product.

Dr. Anurahda Prakki and Dr. Craig Simmons, for acting as members of the research

committee.

Dr. Suja Shrestha, whose technical proficiency and collaboration was indispensible in allowing

the research project to be completed with precision.

My Family, who supported my pursuit of higher education.

Funding provided from the American Association of Endodontists Foundation and the

Canadian Academy of Endodontics Endowment Fund was instrumental in the timely

completion of this project. With sincerity, I extend my appreciation to the work, contribution,

and generosity of these organizations.

 

 

 

 

 

 

 

 

 

 

Page 4: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

 

iv  

Table of Contents

Acknowledgments .......................................................................................................................... iii

Table of Contents ........................................................................................................................... iv

List of Figures ................................................................................................................................ vi

Abbreviations ................................................................................................................................ vii

List of Appendices ....................................................................................................................... viii

Chapter 1 Introduction .................................................................................................................... 1

1.1 General Introduction .......................................................................................................... 1

1.2 Objectives and Hypothesis ................................................................................................ 2

1.2.1 Objectives ................................................................................................................... 2

1.2.2 Hypothesis .................................................................................................................. 2

Chapter 2 Literature Review ........................................................................................................... 3

2.1 Apexification ..................................................................................................................... 3

2.1.1 Calcium Hydroxide ..................................................................................................... 3

2.1.2 Mineral Trioxide Aggregate ....................................................................................... 3

2.2 Regenerative Endodontics ................................................................................................. 4

2.2.1 Cell-Based Approach .................................................................................................. 4

2.2.2 Cell-Free Approach .................................................................................................... 5

2.2.3 Scaffolds ..................................................................................................................... 5

2.3 Bioactive Molecules .......................................................................................................... 6

2.3.1 Transforming Growth Factor β-1 ............................................................................... 7

2.4 Carrier systems .................................................................................................................. 7

2.5 Stem Cells .......................................................................................................................... 8

2.5.1 Stem Cells from Apical Papilla (SCAP) ..................................................................... 9

2.6 Cell Homing ....................................................................................................................... 9

Chapter 3 Article ........................................................................................................................... 11

3.1 Abstract ............................................................................................................................ 11

Page 5: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

 

v  

3.2 Introduction ...................................................................................................................... 12

3.3 Materials and Methods .................................................................................................... 13

3.3.1 Part I – Synthesis and Characterization of Scaffold and TGF-β1-CSnp .................. 14

3.3.2 Part II – Effect of Scaffold Containing TGF-β1-CSnp on SCAP Differentiation .... 18

3.3.3 Statistical Analysis .................................................................................................... 19

3.4 Results .............................................................................................................................. 19

3.4.1 Characterization of Scaffold ..................................................................................... 19

3.4.2 Characterization of TGF-β1-CSnp ........................................................................... 19

3.4.3 Effect of Scaffold Containing TGF-β1-CSnp on SCAP Differentiation .................. 20

3.5 Discussion ........................................................................................................................ 21

3.6 References ........................................................................................................................ 23

Chapter 4 Discussion, Conclusion and Future Directions ............................................................ 27

4.1 General Discussion .......................................................................................................... 27

4.2 Conclusion ....................................................................................................................... 31

4.3 Future Directions ............................................................................................................. 32

References ..................................................................................................................................... 33

Appendix I .................................................................................................................................... 46

Characterization of scaffold.............................................................................................46

Characterization of TGF-β1-CSnp ..................................................................................47

TGF-β1-induced migration of SCAP ..............................................................................48

Effect of nanoparticles on biomineralization ..................................................................48

Immunofluorescence of DMP-1 and DSPP during odontoblast differentiation .............49

 

 

Page 6: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

 

vi  

List of Figures

Figure 1: Characterization of scaffold ..........................................................................................46 (A) Scaffold morphology as observed under SEM illustrating porous structure; (B) Scaffold swelling ratio; (C) Scaffold degradation (%) in the presence of lysozyme; (D) FTIR Spectra of CMCS, Gelatin, and the CMCS/gelatin scaffold; (E) SCAP morphology as observed under SEM illustrating favorable response to scaffold.

Figure 2: Characterization of TGF-β1-CSnp ................................................................................47 (A) Cumulative release (ng) of TGF-β1 from TGF-β1-CSnp; (B) Relative viability of SCAP exposed to Free TGF-β1, TGF-β1-CSnp, or CSnp over 1, 3 and 14 days. (C) Representative fluorescent microscopic view after the calcein-AM staining (upper row, magnification 20X) and phase contrast microscopic images (lower row, magnification 40X). Data represent means ± SD (n = 3). *Significant difference between groups, p < 0.001.

Figure 3: ........................................................................................................................................48 (A) TGF-β1-induced migration of SCAP: Cell migration was examined by a Transwell assay. SCAP were cultured in presence of Free TGF-β1, TGF-β1 released from TGF-β1-CSnp incubated for different time periods and CSnp. The lower membrane surfaces were photographed through a microscope at 20X magnification. Migrated cells in each field were counted. *Significant difference between groups, p < 0.028. (B) Effect of nanoparticles on biomineralization: ARS was quantified for biomineralization in SCAP culture for 3 weeks in presence of Free TGF-β1, TGF-β1-CSnp or CSnp. Data are shown as mean OD/mg of total protein. Data represent means ± SD (n = 3). *Significant difference between groups, p < 0.001.

Figure 4: Immunofluorescence of DMP-1 and DSPP during odontoblast differentiation: ...........49 (A) FITC-conjugated antibody was used to detect the localization of the protein (green signal); all samples were counterstained with DAPI (blue signal). (B) Quantification of protein expression by ImageJ software and data are presented as relative mean fluorescence intensity. *Significant difference between groups, p < 0.001.

 

 

 

Page 7: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

 

vii  

Abbreviations

BM bioactive molecules

CH calcium hydroxide

CMCS carboxymethyl chitosan

CSnp chitosan nanoparticles

DMP-1 dentin matrix protein-1

DPSC dental pulp stem cells

DSPP dentin sialophosphoprotein

MSC mesenchymal stem cells

MTA mineral trioxide aggregate

SCAP stem cells from apical papilla

TGF-β1 transforming growth factor Beta-1

Page 8: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

 

viii  

List of Appendices

 

Appendix I ................................................................................................................................... 46

Page 9: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

1  

Chapter 1 Introduction

1.1 General Introduction Bioactive molecules (BM) are critical to mammalian cell growth (1) and thus are

fundamental in the processes of tissue development, function, and healing. The absence

of these BM, or their presence in inappropriate concentrations in a specific tissue

environment can have diverse effects on cell activity, that results in varying biological

effect on the tissue and highlights the complex multifunctional role that BM play in tissue

homeostasis (2). Understanding BM-cell interactions is central to translational

knowledge in tissue regenerative applications including regenerative endodontics.

Studying how and when these interactions occur on a spatial and temporal basis facilitate

this understanding and expedite the clinical application of this knowledge. Current

regenerative endodontic procedures employ tissue engineering principles that exploit

BM-cell interactions (3).

In the past, and perhaps to some degree in the present, apexification has been the

preferred treatment of immature teeth that have become pulpless. However, immature

teeth, subsequent to successful apexification, remain at risk of fracture due to thin root

dentin walls. Recognizing this, endodontists are developing biologically based treatment

strategies to improve the fate of these often-fractured teeth by generating new functional

tissue that they hope will improve the quality of patient’s lives (4, 5).

An alternative to apexification is a regenerative endodontic procedure. However, at

present the outcomes of these procedures remain questionable (6). Different protocols to

regenerate pulp and dentin have been investigated (7, 8) and different materials and

different BM have been used to stimulate and support new tissue growth (9-13).

Molecules of the transforming growth factor (TGF)-β superfamily are involved in the

regulation of cell proliferation, cell differentiation, epithelial-mesenchymal transition,

and embryonic development (14). Employing tissue-engineering principles, scaffolds,

stem cells and bioactive molecules have been used to regenerate neo-tissue at a desired

site over a predictable period of time. Polymeric-based scaffolds have been investigated

and offer only measured success. Thus, no scaffold material has gained universal

Page 10: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

2  

acceptance. Additionally, augmentation of scaffold bioactivity using BM release to

predictably generate new tissue in dentin pulp engineering has remained a challenge (3).

While no preferred scaffold material exists in regenerative endodontics, the many

positive attributes of a chitosan-based scaffold may make it an optimum material for this

application (15, 16).

Tissue healing and repair of dental pulp and reparative dentin formation, is a result of

distinct BM interaction, which drives cell chemotaxis, proliferation, differentiation, and

extracellular matrix remodeling (17). The current study investigated the potential of a

novel chitosan-based scaffold containing biomolecule (TGF-β1)-releasing CSnp (TGF-

β1-CSnp) to enhance migration and differentiation of SCAP, since cell migration and

differentiation are critical initial steps in the regeneration of the pulp-dentin complex.

1.2 Objectives and Hypothesis

1.2.1 Objectives I. To develop and characterize a chitosan-based scaffold and a novel TGF-β1-releasing

chitosan nanoparticle (CSnp) system.

II. To examine the effect of chitosan scaffold containing TGF-β1-CSnp on migration and

odontogenic differentiation of SCAP.

1.2.2 Hypothesis A bioactive chitosan scaffold containing a sustained TGF-β1-releasing CSnp system will

promote migration and enhance differentiation of SCAP.

Page 11: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

3  

Chapter 2 Literature Review

2

2.1 Apexification Apexification can be defined as a method to induce a calcified barrier at the apical end of

a root with an open apex or continued apical development of an incompletely formed root

in teeth with necrotic pulps (18). Two materials currently used in this method are

calcium hydroxide (CH) and mineral trioxide aggregate (MTA) (19).

2.1.1 Calcium Hydroxide Calcium hydroxide is a low cost, easily obtained, highly alkaline (pH 12) material formed

by the combination of calcium oxide and water. Introduced to dentistry by Hermann (20),

its application on exposed dental pulp leads to the formation of a hard tissue or calcific

bridge over the exposure (21). Similarly, its application within the root canal results in

hard tissue formation in the root canal (pulpotomy) or at the root apex (apexification).

Additional benefits of CH include neutralization of low pH acid, raising of tissue pH,

stimulation of fibroblasts, as well as antimicrobial effect through various mechanisms

such as membrane lipid peroxidation, protein denaturation, and splitting of microbial

DNA strands (21, 22). However, use of CH in apexification has drawbacks. Multiple

appointments over an extended period of time (months to years) are required to disinfect

the root canal and induce the formation of calcified tissue at the root end where it

facilitates the placement of a root canal filling. In addition, prolonged intracanal use of

CH has been shown to alter the dentin matrix, making the tooth susceptible to fracture

(23-26).

2.1.2 Mineral Trioxide Aggregate Mineral trioxide aggregate (MTA) is a powder consisting of tricalcium silicate, tricalcium

aluminate, tricalcium oxide, bismuth oxide, silicate oxide, tetracalciumalumino ferrite,

and calcium sulphate dehydrates (27). When mixed with moisture, these particles set to

form an alkaline (pH 12) colloidal gel initially, which then solidifies to a hard structure

over approximately four hours. MTA is biocompatible and shows excellent sealing

ability when applied to tooth structure. Additionally, new tissue formation has been

Page 12: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

4  

shown in direct apposition to MTA in vivo (28). Due to these favourable characteristics,

it has been employed in immediate or ‘one-step’ apexification procedures. Apically

placed MTA establishes a root-end ‘plug’ by mechanical means against which root filling

material can be packed, while circumventing some of the drawbacks associated with CH

apexification (28, 29). Unfortunately, even when MTA apexification is successful, like

CH, root width is not increased and tooth susceptibility to fracture remains (30).

Children between the ages of 7 to 15 have a high incidence of dental trauma (31). This is

an age when most permanent teeth are incompletely formed and when jaw bone

development makes tooth replacement with an osseo-integrated implant contraindicated

(32, 33). It has also been reported that up to 50% of traumatized teeth may be diagnosed

with pulp necrosis (34). Traumatized teeth with immature roots that are treated

endodontically show a cervical fracture incidence between 28%-77% over four years (35).

Thus, it has been suggested that a treatment approach that increases the tooth root width

and reduces fracture susceptibility, is preferable (36).

2.2 Regenerative Endodontics Regenerative endodontics refers to biologically based procedures designed to

physiologically replace damaged or undeveloped tooth structure, including dentin and

root structures, as well as cells of the pulp-dentin complex (18). These procedures

employ classic tissue engineering techniques comprised of stem cells, scaffolds, and

bioactive growth factors (3). Two such procedures involve cell-based and cell-free

approaches.

2.2.1 Cell-Based Approach A cell-based tissue engineering approach involves the application of cells exogenously to

the site where tissue regeneration is desired. This approach presents several challenges.

Allogeneic cell transplants can undergo immune rejection and lead to heterotopic tissue

formation. Additionally, the sourcing and expansion of cell populations appropriate for

this application can be time-consuming and costly (37). Furthermore, transplanted cells

are particularly vulnerable to their environment, and optimizing the balance between

delivery and consumption of oxygen and nutrients is difficult (38). Selection of cell type

Page 13: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

5  

and concentration to be transplanted needs careful consideration as this has direct impact

on the nutrient availability and metabolism of these cells, which can subsequently affect

the overall performance of the implanted materials (39).

2.2.2 Cell-Free Approach A cell-free tissue engineering approach involves recruitment or homing of endogenous

cells to the desired site of regeneration. This approach may overcome some of the

challenges associated with the cell-based technique previously mentioned (40). Different

protocols for cell-free dentin-pulp tissue regeneration have been studied for many years

(7, 8, 41-43) as have the different materials used to support cell growth and deliver

appropriate bioactive molecules to the site of regeneration (9-13, 15, 16, 44-68).

Employing a conducive scaffold in regenerative endodontic applications is critical.

Several polymeric materials have been used as scaffolds either alone or in combination,

but optimizing a scaffold material together with controlled delivery of bioactive

molecules has remained a challenge.

2.2.3 Scaffolds Scaffolds are materials that serve a structurally supportive role for developing tissue in

regenerative procedures. Additionally, they provide a surface for cell attachment and

may act as a vehicle for bioactive molecule delivery. Materials used for scaffolds in

regenerative applications should be biocompatible, substantive, tunable, porous, reactive,

and biodegradable (3). These materials include natural polymers like polysaccharides

and extra-cellular matrix (ECM); synthetic biodegradable polymers such as polyesters

(e.g. PGA, PLA, PCL) and polyurethanes; bioceramics (e.g. calcium phosphates,

hydroxyapatite, tricalcium phosphate, bioactive glasses); and hydrogels (e.g. collagen,

alginate, chitosan). Blood clots and platelet-rich plasma (PRP) have also been used (69).

2.2.3.1 Chitosan While no ‘gold standard’ scaffold material has been established in regenerative

endodontics, chitosan-based scaffolds may be best suited for dentin-pulp engineering, as

chitosan possesses many desirable attributes for this application. Chitosan is the N-

deacetylated derivative of chitin. Chitin is a naturally abundant polysaccharide, and the

Page 14: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

6  

supporting material of crustaceans and insects; it functions naturally as a structural

polysaccharide (15). Chitosan is a bioactive biopolymer with a cationic charge that offers

substantive benefits when interacting with hard tissues including root dentin (70). It can

be customized to form scaffolds that swell and adapt well to recipient sites (68). It has

alterable porosity to permit transport of nutrients, biomolecules, and toxic cellular

products. Its structural strength and dissolution characteristics can be altered. It is non-

toxic, antibacterial, angiogenic, resistant to endotoxin and inhibits collagen degradation

(16, 55, 68-83). Several studies have reported many favorable characteristics of different

forms of chitosan that match those of an ideal scaffold, highlighting chitosan’s viability

as a scaffold material in tissue regeneration (45, 68, 70, 73, 76, 78-82). Carboxymethyl

chitosan (CMCS), a water-soluble derivative of chitosan, has displayed several beneficial

characteristics when used as a scaffold (84), and as a surface-modifier of dentin matrix to

enhance antibacterial efficacy and ultrastructural characteristics (83, 85).

2.2.3.2 Gelatin Gelatin is a natural polymer obtained by a controlled acid or alkaline hydrolysis of native

collagen (86). Due to its excellent biocompatibility, biodegradability, and nontoxicity,

gelatin has been employed in medicine and pharmaceutics as a material for controlled

drug release (87). Gelatin alone does not offer appropriate strength or substantivity when

used in tissue regenerative applications. However, when combined with chitosan, gelatin

produces improved mechanical properties and swelling capacity of the scaffolds in an

aqueous environment (88).

2.3 Bioactive Molecules Bioactive molecules is an umbrella term referring to a diverse group of molecules

encompassing growth factors, chemokines, cytokines, extracellular matrix molecules, and

bioactive peptides. Growth factors can be defined as extracellular signalling proteins that

are involved in cell to cell communications (89). Some concerns regarding the

application of BM in regenerative procedures include the potential for their uncontrolled

release, toxicity, short half-life, and possible lack of effect. The release of BM from

polymeric materials can occur over an extended period of time without loss of bioactivity

(90-92). The culture time for complete osteogenic differentiation of mesenchymal stem

Page 15: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

7  

cells in vitro has been reported to take place over 3 weeks, but this process occurs faster

in the presence of certain BM (92-95), highlighting the importance of evaluating the

beneficial effects of sustained release of BM in this study. One of the most common

strategies to achieve sustained release of BM is to incorporate them into polymeric

biomaterials.

2.3.1 Transforming Growth Factor β-1 Several BM have been evaluated in tissue engineering (96, 97) and many different BM

have been reported to be sequestered within the dentin matrix (98). The current literature

indicates that the transforming growth factor (TGF)- β superfamily is one of the most

important growth factors in dentin-pulp regeneration (99, 100). The TGF-β superfamily

regulates cell proliferation, cell differentiation, the epithelial-mesenchymal transition and

embryonic development (14). An in vitro human tooth model based investigation

suggested TGF-β1 to have direct involvement in the regulation of cell proliferation,

migration and extracellular matrix synthesis in the human dental pulp and in the repair

process occurring after tooth injury (101). It has been shown that TGF-β1 localizes in the

dentin matrix, and implantation of isolated dentin proteins and dentin matrix (102, 103)

in dental pulp induced odontoblast-like differentiation. TGF-β1 cell signalling may occur

through TGF-β receptor internalization into the cell via clathrin-coated vesicle

endocytosis as well as via membrane caveolae (104). Clathrin-mediated endocytosis has

been shown to promote TGF-β-induced Smad activation and transcriptional responses.

Caveolae are regarded as signaling centers for G protein-coupled receptors and tyrosine

kinase receptors, but they are indicated to facilitate the degradation of TGF-β receptors

and therefore, turnoff of TGF-β signaling (104). Evidence also indicates that TGF-β

signaling can take place on the cell plasma membrane (105).

2.4 Carrier systems Therapeutic applications of growth factors pose a major challenge clinically due to their

short half-life and rapid diffusion into the surrounding medium and this may have

deleterious effects in vivo (106). Bioactive molecules can be introduced into many tissue

engineering systems by various methods, which include adding directly to the medium,

by genetically engineering cells to overexpress them, and by constructing polymeric

Page 16: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

8  

systems that allow their controlled release (107). Challenges of biomolecule delivery

previously mentioned may be overcome if polymeric carrier systems are engineered to

deliver bioactive molecules over time and with a degree of spatial control. This provides

an effective concentration of active BM for endogenous cells since spatiotemporal

expression of BM has been shown in odontogenesis in vivo (108). Examples of BM

carriers include the scaffold itself, microparticles, or nanoparticles. Khil et al. (109)

designed a porous chitosan scaffold, containing chitosan microspheres loaded with TGF-

β1, to enhance chondrogenesis. They demonstrated that the scaffolds containing the

loaded chitosan microspheres significantly increased the cell proliferation and production

of extracellular matrix (ECM). A similar approach using chitosan-based materials has

been reported (107), where three dimensional collagen/chitosan/glycosaminoglycan

scaffolds were seeded with rabbit chondrocytes and combined with TGF-β1-loaded

chitosan microspheres. This allowed for an evaluation of the effect of released TGF-β1

on the chondrogenic potential of rabbit chondrocytes in such a system. Nanoparticles are

very small particles, approximately 1-100nm in size (110). Functionalized chitosan

nanoparticles have been shown to be effective in the temporal-controlled delivery of BM

to an odontogenic line of stem cells (111, 112) and its use has enhanced cell proliferation

and differentiation in dentin-pulp engineering (113, 114).

2.5 Stem Cells Stem cells are unspecialized, immature cells with the potential to develop into many

different cell lineages via differentiation and hence they are useful in regenerative

strategies (115). The Mesenchymal and Tissue Stem Cell Committee of the International

Society for Cellular Therapy (ISCT) proposed minimal criteria to define human MSC

(116). First, MSC must be plastic-adherent when maintained in standard culture

conditions. Second, MSC must express CD105, CD73 and CD90, and lack expression of

CD45, CD34, and other negative markers. Third, MSC must differentiate to osteoblasts,

adipocytes and chondroblasts in vitro. Various tissue-specific stem cell populations have

been described including those pertaining to oral tissues (117, 118). Some of these stem

cells include salivary gland stem cells (SGSC), oral epithelial stem cells (OESC),

periosteal derived stem cells (PSC), dental follicle progenitor cells (DFPC), dental pulp

Page 17: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

9  

stem cells (DPSC), stem cells from human exfoliating deciduous teeth (SHED),

periodontal ligament stem cells (PDLSC), inflammatory periapical progenitor cells

(IPPC), bone marrow stem cells (BMSC), and stem cells from the apical papilla (SCAP)

(119).

2.5.1 Stem Cells from Apical Papilla (SCAP) Stem cells from the apical papilla possess several characteristics that make them an

excellent choice for studying dentin-pulp tissue regeneration. First characterized in 2006,

SCAP are mesenchymal stem cells in the apical papilla of developing teeth that can

differentiate into odontoblast-like cells forming dentin (120). It has been shown that

SCAP play a role in root development (121). Furthermore, SCAP have demonstrated an

elevated tissue regeneration capacity, higher telomerase activity, and improved migration

capacity when compared to DPSC from the same tooth (120). SCAP have been shown to

regenerate vascularized human dental pulp-like tissues suggesting their potential as a

source of primary odontoblasts (122). Previous investigators have demonstrated that

SCAP show a significantly higher proliferation rate and mineralization potential than

DPSC (123). In addition, SCAP are derived from a very accessible tissue resource and

are relatively easy to expand in vitro so as to provide enough cells for potential clinical

applications. Isolated SCAP grown in cultures can undergo dentinogenic differentiation

when stimulated with biomolecules and have also been shown to play a potential role in

continued root formation of immature pulpless teeth (62). Studies have shown that SCAP

cells remain viable and proliferate after exposure of dentin to EDTA, NaOCl, and CH,

which are typical irrigants and medicaments used in endodontics (124, 125).

2.6 Cell Homing Cell homing has been regarded as a process by which hematopoietic stem cells exit from

blood vessels (trans-endothelialization) and migrate to a site (126). In tissue engineering,

cell homing is the active recruitment of endogenous cells, including stem/progenitor cells

into the anatomic site of regeneration. Cell homing techniques have been used in the

regeneration of dental tissues (127, 128). Chemotaxis-induced cell homing has been

shown to result in re-cellularization and revascularization in the endodontically prepared

root canal in vivo (97, 129). Platelet rich plasma (PRP) has been a popular focus of

Page 18: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

10  

investigation as a scaffold as well as a homing tool due to its rich supply of autologous

BM (13, 60, 130-133). However, PRP preparation is cumbersome and control of platelet

concentration and therefore control of BM concentration is unpredictable and may lead to

a detrimental effect on stem cells (134). Since, as mentioned, BM have been shown to be

effective in recruiting and encouraging odontogenic differentiation of stem/progenitor

cells, the use of specific BM may prove to be essential to the successful evolution of

regenerative protocols in the management of immature permanent teeth with necrotic

pulps.

 

 

 

   

 

   

   

 

   

 

 

Page 19: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

11  

Chapter 3 Article

3

3.1 Abstract Introduction: This two-part study hypothesized that novel modified chitosan-based

scaffold containing a sustained transforming growth factor (TGF)-β1-releasing

nanoparticle system (TGF-β1-CSnp) will promote migration and enhance differentiation

of stem cells from the apical papilla (SCAP) as compared to Free TGF -β1 + scaffold,

and the scaffold alone. Methods: Part I describes the synthesis and characterization of a

carboxymethyl chitosan (CMCS) based scaffold and the characterization of the TGF-β1-

CSnp. Part II examines the effect of sustained TGF-β1 release from scaffold containing

TGF-β1-CSnp on SCAP vitality, migration, and odontogenic differentiation. Results:

The scaffold demonstrated acceptable biocompatibility and physical properties conducive

to the maintenance of cellular function. The incorporation of TGF-β1 in CSnp allowed

for sustained release of a critical concentration of TGF-β1 to SCAP over the term of the

study. Except for an initial period of increased SCAP migration noted with Free TGF-β1

+ scaffold, SCAP showed greater viability, migration, biomineralization, and

odontogenic potential, with TGF-β1-CSnp + scaffold than with Free TGF-β1 + scaffold

or CSnp + scaffold. Conclusions: These experiments highlighted the potential of a

CMCS based scaffold with engineered growth factor releasing nanoparticles to promote

migration and differentiation of SCAP over time, and may have direct application to

improve current endodontic regenerative protocols.

Key words: Stem cells from apical papilla, chitosan nanoparticles, transforming growth

factor-β1, cell migration, odontogenic differentiation, regenerative endodontic procedures

Page 20: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

12  

 

3.2 Introduction Tooth retention has mechanical, biological, and psychological benefits that include

chewing efficiency, speech, immune-competence, and self-confidence (4, 5). Endodontic

treatment of mature permanent teeth with apical periodontitis has proven to be highly

successful in retaining teeth that at one time were routinely extracted (135). Treatment of

immature permanent teeth with apical periodontitis has proven to be more challenging

and less predictable. Currently, the treatment strategies used in the management of

immature teeth with necrotic pulps include apexification using calcium hydroxide,

placement of an apical ‘plug’ of MTA, and regenerative endodontic procedures.

Apexification requires multiple appointments over an extended period of time to disinfect

the root canal, and induce formation of calcified tissue at the root end to facilitate

placement of a root canal filling. An apically placed MTA ‘plug’ attempts to achieve the

same end mechanically but usually requires less treatment appointments. However both

of these treatment strategies fail to produce an increase in the thickness of the root canal

wall even when successful, leaving the root susceptible to fracture over time (30).

Regenerative endodontics is a viable treatment alternative, but the predictability for a

desirable treatment outcome remains questionable (6). Different protocols to regenerate

pulp and dentin have been investigated (7, 8) and different materials and different

bioactive molecules (BM) have been used to stimulate and support new tissue growth (9-

13). The current literature indicates that members of the transforming growth factor

(TGF)- β superfamily are important growth factors in dentin-pulp regeneration (99, 100)

and have played a role in the regulation of cell proliferation, cell differentiation,

epithelial-mesenchymal transition, and embryonic development (14). A scaffold-based

regenerative protocol uses an engineered substrate to attract a stem cell population to a

specific site. In regenerative endodontics, the scaffold should ideally possess tissue-

forming qualities that are expressed in suitable concentrations at opportune times.

Several polymeric-based scaffolds have been used in regenerative endodontics with

measured success, however, optimizing their bioactivity with temporal-controlled BM to

enhance new tissue formation has remained a challenge (3). While no preferred scaffold

Page 21: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

13  

material exists, a chitosan-based scaffold may prove to be preferable since chitosan

exhibits many properties favorable to new pulp and dentin formation.

Chitosan is a ubiquitous bioactive biopolymer with a cationic charge that offers

substantive benefits when interacting with hard tissues including root dentin (70). It can

be customized to form scaffolds that swell and adapt well to recipient sites (68) and it has

alterable porosity to permit transport of nutrients, biomolecules, and toxic cellular

products. Its structural strength and dissolution characteristics can be altered, and it is

non-toxic, antibacterial, angiogenic, and resistant to endotoxin and bacterial degradation

(16, 70, 83).

Carboxymethyl chitosan (CMCS), a water-soluble derivative of chitosan, has displayed

several favorable characteristics when used as a scaffold (84), and as a surface-modifier

of dentin matrix since it has the ability to enhance antibacterial efficacy and stabilize the

dentin matrix (83, 85). Functionalized CSnp have shown effectiveness in the temporal

(time)-controlled delivery of BM to an odontogenic line of stem cells (111, 112), and the

ability to enhance cell proliferation and differentiation in tissue regeneration (113, 114).

Since cell migration and differentiation is a critical initial step in the regeneration of pulp

and dentin, the current study investigated the potential of a novel CMCS scaffold

containing TGF-β1-releasing CSnp (TGF-β1-CSnp) to enhance migration and

differentiation of SCAP.

3.3 Materials and Methods In the first part of this study we assessed the physical and chemical characteristics and

biocompatibility of a synthesized CMCS-based scaffold and TGF-β1-CSnp. In the

second part we assessed the effect of sustained release of TGF-β1 from the scaffold

containing TGF-β1-CSnp on SCAP migration and odontogenic differentiation relative to

the effect of Free TGF-β1 + scaffold, unloaded chitosan nanoparticles + scaffold and the

scaffold alone. All chemicals used in the study were of analytical grade and purchased

from Sigma-Aldrich Inc. (St Louis, MO) unless noted otherwise. TGF-β1 (catalog #

ab50036) and the corresponding ELISA kit were purchased from Abcam Inc. (Toronto,

ON, Canada).

Page 22: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

14  

3.3.1 Part I – Synthesis and Characterization of Scaffold and TGF-β1-CSnp

3.3.1.1 Synthesis and Characterization of Scaffold To prepare the scaffold a 2% (1.5 mL) CMCS solution was added to 15% (1.5 mL) warm

gelatin solution and mixed in a 35 mm tissue culture plate to produce a gel. The gel was

kept at -20°C overnight, and lyophilized at -60°C for 24 h. The dry scaffolds were

immersed in a 2 mL solution of 40% ethanol containing 50 mM methane sulfonic acid

(MES) (pH 5.0), 33 mM N-(3-dimethylaminopropyl)-N’-ethylcarbodiimide hydrochloride

(EDC) and 8 mM N-hydroxysuccinimide (NHS) for 10 h at room temperature. After

neutralizing with 0.1 M Na2HPO4 (pH 9.1) for 1 h, the scaffolds were washed and

lyophilized again for 24 h before testing.

3.3.1.1.1 Porosity Analysis Three scaffolds in each group to be tested later were weighed before and after immersion

in absolute ethanol overnight. Porosity of the scaffolds was calculated using the

following formula (136);

Interval porosity (%) = (!  !  !!)!!!!!!(!!!!!)!!  

 ×  100

Where, 𝑊 = wet weight, 𝑊! = dry weight, 𝜌!= average density of CMCS and gelatin

(0.95 g/mL), 𝜌! = density of anhydrous alcohol (0.79 g/mL).

3.3.1.1.2 Swelling Analysis Dry weight of each scaffold was recorded before and after immersion in PBS at 37°C.

Wet scaffolds were removed at regular time intervals, gently blotted with filter paper and

weighed. The following formula was used to calculate the swelling ratio (137);

Swelling ratio (%) = !!"#!  !!"#

!!"#  ×  100

Where, 𝑊!"# = initial dry weight, 𝑊!"#= weight after swelling

Page 23: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

15  

3.3.1.1.3 Biodegradation Analysis Scaffolds (20 ± 1 mg) were sterilized in 100% ethanol overnight and washed with PBS.

Sterilized scaffolds were immersed in 2 mL of PBS containing 10 mg/L lysozyme (Fisher

Bioreagents; catalog # BP535-1; Activity = 24,200 units/mg), and incubated at 37°C.

Lysozyme was refreshed every 3 days. Scaffolds were removed from the lysozyme

solution at different time intervals, washed, kept at -20°C for 2 h, lyophilized, then

measured to determine dry weight. Degree of degradation was calculated using the

following formula;

Degradation (%) = !!!  !!

!!  ×  100

Where, 𝑊! = initial dry weight, 𝑊! = dry weight after degradation

3.3.1.1.4 Chemical Characterization Chemical characterization of the scaffold was carried out by Fourier transform infrared

(FTIR) spectroscopy. Transmission spectra of powdered scaffold mixed with potassium

bromide in a 1:10 ratio (w/w) were recorded using a FTIR Spectrophotometer (Paragon

500, Perkin Elmer, Waltham, MA, USA) from 4000 to 400 cm-1 with 16 scans and 4 cm-1

resolution.

3.3.1.1.5 SCAP Viability in Scaffold A characterized SCAP cell line was used in the study (138). Cells were cultured in α-

minimum essential medium (MEM) supplemented with 10% fetal bovine serum, 2 mM

L-glutamine and 100-units/mL antibiotic. Sub-confluent SCAP of passage 3 were

detached using trypsin/EDTA and re-suspended at an appropriate concentration. Cultures

were kept at 37°C in a humidified incubator with an atmosphere of 5% CO2. (Thermo

Scientific, Waltham, MA, USA). Dry scaffolds (4 mm diameter) were immersed in

100% ethanol overnight in 96-well plates, washed with PBS, then immersed in culture

medium overnight. SCAP (2X104) were cultured in the scaffolds for 1 week. SCAP

viability was quantitatively analyzed using a Live/Dead viability/cytotoxicity assay kit

(Invitrogen, Carlsbad, CA). Indicator dyes (1µM Calcein-AM and 2µM EthD-1 in PBS,

Molecular Probes, Inc. Eugene, OR, USA) were added and the fluorescence signal

(excitation: 485/530 nm; emission: 530/645 nm) from the resulting solution was obtained.

Page 24: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

16  

The background signals (scaffolds without cells) were subtracted before calculating the

cell viability. Subsequently, scaffolds were fixed overnight with 2.5% glutaraldehyde

(4°C) and dehydrated in graded ethanol and hexamethylenedisilane for scanning electron

microscopy (Hitachi S-2500, Japan).

3.3.1.2 Synthesis and Characterization of TGF-β1-CSnp TGF-β1-CSnp was synthesized by the adsorption method previously described (113).

Briefly, freshly prepared CSnp was re-suspended in 10 mL PBS, to which 2 µg of TGF-

β1 dissolved in 400 µL of 4 mM HCl was added. The mixture was stirred at 4°C

overnight. Nanoparticles were washed with isopropyl alcohol, then water and lyophilized

before use.

3.3.1.2.1 Particle Size and Charge The size/charge of nanoparticles was determined by transmission electron microscopy

(CM12 Philips, The Netherlands) and dynamic light scattering using a Zetasizer (Nano

series, Nano-ZS90, Malvern, UK), respectively.

3.3.1.2.2 Release Kinetics Release kinetics of TGF-β1-CSnp were studied, following manufacturer instructions in

the ELISA kit. Briefly, the amount of TGF-β1 released by 10 mg of TGF-β1-CSnp in 1

mL PBS (pH 7.4) at 37°C was measured by analyzing the supernatant of the

centrifuged mixture. Samples were centrifuged, and at predetermined time points, the

supernatant was collected and immediately frozen. Frozen supernatant was then allowed

to thaw and the amount of TGF-β1 in the mixture was quantified.

3.3.1.2.3 SCAP Viability SCAP viability was quantitatively analyzed with a resazurin-based assay using a viability

assay kit (Promega, USA). Approximately 2X104 SCAP were seeded into 96-well plates

and incubated at 37°C for 24 h. Cells were treated with a nanoparticle suspension (300

µg/mL) or Free TGF-β1 (5 ng/mL) in a culture medium and cultured for 14 days. SCAP

cultured in the absence of nanoparticles or BM was used as a control. Groups tested were:

(a) no nanoparticles, (b) Free TGF-β1, (c) TGF-β1-CSnp and (d) CSnp. Cell survival was

determined by adding 40 µL of reagent directly into the cell culture. Fluorescence was

Page 25: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

17  

measured after incubation (37°C) for 4 h. SCAP stained with calcein-AM was examined

by fluorescent microscopy (Vert.A1, Carl Zeiss, Germany) and phase contrast

microscopy to assess cell morphology.

3.3.1.2.4 Migration Assay SCAP migration was assessed using a 24-well Transwell migration assay (6.5 mm

diameter; 8µm pore size polycarbonate membrane, Corning, Lowell, MA). SCAP

(1X104) in supplement free α-MEM were seeded into the upper chambers and allowed to

attach for 4 h before exposure to test conditions. The lower chambers of the multi-well

system contained test solutions conditioned with (a) no nanoparticles, (b) Free TGF-β1

(10 ng/mL) + scaffold, (c) TGF-β1-CSnp (2 mg/mL) + scaffold or (d) CSnp (2 mg/mL) +

scaffold for pre-determined time points. Cells were cultured at 37°C in 5% CO2. After

incubation for 24 h, cells were fixed in 4% paraformaldehyde for 10 min, and incubated

with 25 µg/ml 4’,6-diamidino-2-phenylindole (DAPI) for 10 min at room temperature.

Non-migrated cells on the upper surface of the membrane were carefully removed and

cells that had migrated through the membrane to the lower surface of the well were

imaged using a fluorescence microscope. Migrated cells from 14 fields taken from 3

wells of each group were counted to assess cell migration.

3.3.1.2.5 Biomineralization Assay SCAP (5X104) were grown to confluence in 6-well culture plates in standard culture

medium. Standard medium was then replaced with a mineralizing medium containing 50

µg/mL ascorbic acid, 10 mM β-glycerolphosphate and 1.8 mM KH2PO4 in standard

medium (113). Free TGF-β1 (3 ng/mL), TGF-β1-CSnp (500 µg/mL) or CSnp (500

µg/mL) was then added to the mineralizing medium and SCAP for 21 days at 37°C and

5% CO2. Biomineralization potential was assessed by staining with alizarin red stain

(ARS). In brief, cells were washed 3 times with PBS and fixed in 10% normal buffered

formalin for 30 min at room temperature. After additional washing, they were incubated

in 2% ARS (pH 4.2) for 20 min at room temperature with gentle agitation, followed by

washing and air-drying. Stained cells were incubated with a 10% cetylpyridinium

chloride in 10 mM sodium phosphate buffer for 30 min to elute calcium-bound stain.

Supernatant was collected, and the optical density determined at 560 nm. Mineralized

Page 26: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

18  

nodule formation was represented as OD per mg of total cellular protein, determined by a

BCA protein assay kit (Sigma-Aldrich, St Louis, MO, USA). Background staining caused

by CSnp without cells was subtracted from the experimental data to provide a net score.

3.3.2 Part II – Effect of Scaffold Containing TGF-β1-CSnp on SCAP Differentiation

3.3.2.1 Preparation of Scaffold Containing Nanoparticles Solution containing Free TGF-β1 (9 ng), TGF-β1-CSnp (1.5 mg) or CSnp (1.5 mg) in

500 µL ethanol was carefully introduced into scaffold 35 mm in diameter and

immediately lyophilized to prepare the scaffolds containing nanoparticles for immediate

use (107).

3.3.2.2 Immunofluorescence Analysis Scaffolds containing Free TGF-β1, TGF-β1-CSnp or CSnp were placed in a 6-well plate

and seeded with SCAP (5X104 cells/well) in mineralizing medium. Test groups included

(a) Free TGF-β1 + scaffold, (b) TGF-β1-CSnp + scaffold, (c) CSnp + scaffold and (d)

scaffold. After 14 d of culture at 37°C, a 20 mm diameter sample from each group was

frozen in Tissue-Tek O.C.T. Compound (Electron Microscopy Sciences, Hatfield, USA)

in liquid nitrogen and sectioned at 25-30 mm in a cryostat at -20°C. Sections were fixed

in 4% paraformaldehyde containing 0.1% Triton-X 100 at 4°C for 30 min, then washed

and blocked with 2.5% BSA for 30 min. Sections were then incubated with mouse anti-

DMP-1 antibody (catalog# sc-73633; Santa Cruz Biotechnologies, CA, USA) and mouse

anti-DSPP antibody (catalog# sc-73632; Santa Cruz Biotechnologies, CA, USA)

antibody, diluted 1:50 in blocking reagent at 37°C for 2 h. After several washes in PBS

/Tween 20, specimens were incubated with secondary antibody from goat anti-mouse IgG

FITC conjugate (catalog# sc-2010; Santa Cruz Biotechnologies, CA, USA) and diluted

1:1500 in PBS at 37°C for 1 h. After rinsing, specimens were counterstained in DAPI and

examined under confocal laser scanning microscopy (Leica Microsystems, Illinois, USA).

Quantitation of proteins expression was carried out using ImageJ software (NIH,

Bethesda, MA, USA) to calculate relative mean fluorescence intensity.

Page 27: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

19  

3.3.3 Statistical Analysis Data obtained from this study are expressed as mean ± standard deviation. Statistical

analyses were performed using one-way analysis of variance (ANOVA) and post hoc

Tukey test. A P value <0.05 was considered statistically significant.

3.4 Results

3.4.1 Characterization of Scaffold

3.4.1.1 Physicochemical Properties: The ultrastructure of the scaffold showed porous structure (Fig. 1A), with an interval

porosity of 78.3 ± 1.8%. The swelling ratio was calculated to be 763.5 ± 164% (by

weight) after 24 h (Fig. 1B). Degradation study showed that 40 ± 8.4% of the scaffold

was degraded in 3 weeks (Fig. 1C). FTIR spectra displayed presence of cross-linked

CMCS and gelatin in the scaffold (Fig. 1D). The band corresponding to N-H stretching

had shifted to a lower wavelength (3270 cm-1), when compared to CMCS (3350 cm-1)

and gelatin (3396 cm-1), and was slightly broadened. The broadening of the band at 3270

cm-1 of the cross-linked samples was due to OH vibration (H-bonded/non-bonded) from

the CMCS molecules. The formation of amide linkage between amino groups of CMCS

and carboxyl groups of gelatin was confirmed by the shift of the amide II (1514 and 1426

cm-1) and carbonyl bands (1628 cm-1). The band at 1232 cm-1 corresponded to amide III.

The absorption band at 1331 cm-1 was thought to be due to symmetric stretching

vibration of –COOH in the CMCS. Bands at 1110 cm-1 and at 1054 cm-1were

characteristic for -C-O- and –C-O-C- in the CMCS.

3.4.1.2 Effect of Scaffold on SCAP Viability Relative viability of SCAP in the presence of scaffold was 81.2 ± 2% at day 1 and 84.4%

at day 7. SCAP morphology appeared normal under SEM (Fig. 1E).

3.4.2 Characterization of TGF-β1-CSnp

3.4.2.1 Physicochemical Properties Size of TGF-β1-CSnp was 58.9±12 nm and charge was +19.1±1.4. Figure 2A shows the

cumulative release (ng) of TGF-β1 from TGF-β1-CSnp in a sustained manner for up to

Page 28: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

20  

28 days.

3.4.2.2 Effect on SCAP Viability SCAP were viable in the presence of TGF-β1-CSnp as shown in Figure 2B. A

statistically significant (p<0.001) increase in viability was shown at days 3 and 14 in the

TGF-β1-CSnp group. Viability assessment was further supported by cell morphology as

observed by phase contrast microscopy (Fig. 2C).

3.4.2.3 Effect on SCAP Migration At day 1 SCAP migration with Free TGF-β1 + scaffold was significantly higher

(p<0.001) than that seen with TGF-β1-CSnp + scaffold (Fig. 3A). At day 7 cell

migration with Free TGF-β1 + scaffold was significantly reduced by 80% (p<0.001) and

showed only a 10% increase after 14 and 21 days. There was no statistical difference in

cell migration in the TGF-β1-CSnp + scaffold group between 1 and 7 days, and a 10%

increase in cell migration after 14 and 21 days as compared to that seen at 7 days. There

was a statistically significant difference (p<0.028) in cell migration between Free TGF-

β1 + scaffold and TGF-β1-CSnp + scaffold group after 21 days.

3.4.2.4 Effect on Biomineralization Biomineralization was significantly higher (p<0.001) in the TGF-β1-CSnp + scaffold

group after 7 days than that seen in Free TGF-β1 + scaffold and CSnp + scaffold groups

(Fig. 3B). Biomineralization was also higher in the TGF-β1-CSnp + scaffold group than

the Free TGF-β1 + scaffold or CSnp + scaffold groups after 14 day. No significant

difference between groups was seen at 21 days.

3.4.3 Effect of Scaffold Containing TGF-β1-CSnp on SCAP Differentiation

3.4.3.1 Immunofluorescence analysis Signals for DMP-1 and DSPP were significantly (p<0.001) higher in the TGF-β1-CSnp +

scaffold group than in the Free TGF-β1 + scaffold group (Fig. 4A and 4B). There were

markedly lower signals for both DMP-1 and DSPP in CSnp + scaffold and scaffold only

groups (Fig. 4A and 4B). Immunofluorescence analysis indicated a higher level of SCAP

Page 29: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

21  

differentiation in the scaffold and a more uniform distribution of the cells in TGF-β1-

CSnp + scaffold group, when compared to other groups.

3.5 Discussion A scaffold capable of attracting stem cells and delivering BM in a temporal-controlled

manner is essential for optimizing tissue engineering. In this study, we described a

CMCS scaffold containing controlled TGF-β1-releasing CSnp that may have practical

application to regenerative endodontics. The engineered matrix/nanoparticle scaffold

mimicked dental extracellular matrix, possessed high porosity, and significantly

enhanced SCAP migration and differentiation.

Scaffolds fabricated by cross-linking gelatin with chitosan derivatives produced

improved mechanical properties and increased swelling capacity (137). Increases in

swelling capacity of the scaffold may be desirable for endodontic applications to improve

scaffold substantivity and adaptation to the root canal walls after implantation.

Favorable scaffold-cell interaction, as observed through SEM in our study, infers that the

CMCS scaffold used in this study was compatible with SCAP. SCAP are ecto-

mesenchymal stem cells from the apical papilla of developing human teeth and represent

a population of cells that can migrate to the root canal wall and participate in tissue

regeneration or organized repair. SCAP has demonstrated increased migration, tissue

regeneration capacity, and telomerase activity, when compared to dental pulp stem cells

from the same tooth (120). It has also been suggested that SCAP may be better equipped

to survive and maintain their potential for differentiation in adverse, low oxygen

conditions (119).

TGF-β1 is a BM released from cells and incorporated into dentin and other non-

collagenous proteins. It has been identified as a promoter of odontogenic differentiation

(103). However, availability of this molecule and other BM sequestered in dentin matrix

may not present in an optimum concentration or for a duration necessary to satisfy stem

cell requirements in tissue regeneration (139). In the environment of a pulpless root

canal, it is crucial that timely release of an optimum concentration of growth factors be

available for cells to survive, migrate and differentiate (140). The importance of sustained

Page 30: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

22  

release of BM for cell migration and differentiation has previously been shown (113).

Different concentrations of TGF-β1 were employed in this study since it has been shown

that TGF-β1 can effectively promote or inhibit cell activity depending on the available

concentration (107, 141). The significant increase in SCAP viability compared to the

same concentration of non-loaded CSnp or Free TGF-β1 was attributed to the release of

bioactive TGF-β1 in a controlled manner over time. The potential to provide such an

environment was demonstrated in the TGF-β1-CSnp + scaffold group where SCAP

appeared to survive, migrate, and differentiate better than in the other treatment groups.

TGF-β1 is known to modulate many cell activities including cell migration (140).

Significantly higher cell migration after 1 day in the Free TGF-β1 + scaffold group

compared with the TGF-β1-CSnp + scaffold group was expected since all of the TGF-β1

became available to the cells immediately when the TGF-β1 was in free form. After day

7, cell migration in the Free TGF-β1 + scaffold group decreased significantly, which

reflected a significant loss of bioactivity. In contrast, sustained release of TGF-β1 from

TGF-β1-CSnp + scaffold group led to sustained cell migration over time. This was an

important finding in our study, as it showed the ability of the bioactive scaffold

containing TGF-β1-CSnp to address the issue of the short half-life of TGF-β1 (two to

three minutes) when it is used in free form in regenerative procedures (142).

Enhanced biomineralization of SCAP in the presence of TGF-β1-CSnp + scaffold was

also seen and can be attributed to the controlled release of TGF-β1 during the longer

periods of observation, as were the higher levels of DMP-1 and DSPP expression. The

immunofluorescent images also demonstrated more uniform distribution of the cells in

the TGF-β1-CSnp + scaffold group, when compared to other experimental groups

indicating that TGF-β1 release homogenously from the CSnp dispersed in the scaffold.

Page 31: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

23  

3.6 References 1. Fiske J, Davis DM, Leung KC, McMillan AS, Scott BJ. The emotional effects of

tooth loss in partially dentate people attending prosthodontic clinics in dental schools in

England, Scotland and Hong Kong: a preliminary investigation. Int Dent J.

2001;51(6):457-62.

2. Bergenholtz G. Effect of bacterial products on inflammatory reactions in the dental

pulp. Scand J Dent Res. 1977;85(2):122-9.

3. de Chevigny C, Dao TT, Basrani BR, Marquis V, Farzaneh M, Abitbol S, et al.

Treatment outcome in endodontics: the Toronto study--phase 4: initial treatment. J

Endod. 2008;34(3):258-63.

4. Jeeruphan T, Jantarat J, Yanpiset K, Suwannapan L, Khewsawai P, Hargreaves KM.

Mahidol study 1: comparison of radiographic and survival outcomes of immature teeth

treated with either regenerative endodontic or apexification methods: a retrospective

study. J Endod. 2012;38(10):1330-6.

5. Lee BN, Moon JW, Chang HS, Hwang IN, Oh WM, Hwang YC. A review of the

regenerative endodontic treatment procedure. Restor Dent Endod. 2015;40(3):179-87.

6. Hargreaves KM, Diogenes A, Teixeira FB. Treatment options: biological basis of

regenerative endodontic procedures. J Endod. 2013;39(3 Suppl):S30-43.

7. Bose R, Nummikoski P, Hargreaves K. A retrospective evaluation of radiographic

outcomes in immature teeth with necrotic root canal systems treated with regenerative

endodontic procedures. J Endod. 2009;35(10):1343-9.

8. Ostby BN. The role of the blood clot in endodontic therapy. An experimental

histologic study. Acta Odontol Scand. 1961;19:324-53.

9. Nygaard-Ostby B, Hjortdal O. Tissue formation in the root canal following pulp

removal. Scand J Dent Res. 1971;79(5):333-49.

10. Vacanti JP, Morse MA, Saltzman WM, Domb AJ, Perez-Atayde A, Langer R.

Selective cell transplantation using bioabsorbable artificial polymers as matrices. J

Pediatr Surg. 1988;23(1 Pt 2):3-9.

11. Leach JB, Schmidt CE. Characterization of protein release from photocrosslinkable

hyaluronic acid-polyethylene glycol hydrogel tissue engineering scaffolds. Biomaterials.

2005;26(2):125-35.

Page 32: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

24  

12. Martin G, Ricucci D, Gibbs JL, Lin LM. Histological findings of

revascularized/revitalized immature permanent molar with apical periodontitis using

platelet-rich plasma. J Endod. 2013;39(1):138-44.

13. Nakashima M, Reddi AH. The application of bone morphogenetic proteins to dental

tissue engineering. Nat Biotechnol. 2003;21(9):1025-32.

14. Bashutski JD, Wang HL. Role of platelet-rich plasma in soft tissue root-coverage

procedures: a review. Quintessence Int. 2008;39(6):473-83.

15. Lee DS, Yoon WJ, Cho ES, Kim HJ, Gronostajski RM, Cho MI, et al. Crosstalk

between nuclear factor I-C and transforming growth factor-β1 signaling regulates

odontoblast differentiation and homeostasis. PLoS One. 2011;6(12):e29160.

16. Chan BP, Leong KW. Scaffolding in tissue engineering: general approaches and

tissue-specific considerations. Eur Spine J. 2008;17(Suppl 4):467-79.

17. Shrestha A, Friedman S, Kishen A. Photodynamically crosslinked and chitosan-

incorporated dentin collagen. J Dent Res. 2011;90(11):1346-51.

18. Bernkop-Schnurch A. Chitosan and its derivatives: potential excipients for peroral

peptide delivery systems. Int J Pharm. 2000;194(1):1-13.

19. Kumar MN, Muzzarelli RA, Muzzarelli C, Sashiwa H, Domb AJ. Chitosan chemistry

and pharmaceutical perspectives. Chem Rev. 2004;104(12):6017-84.

20. Kishen A, Shi Z, Shrestha A, Neoh KG. An investigation on the antibacterial and

antibiofilm efficacy of cationic nanoparticulates for root canal disinfection. J Endod.

2008;34(12):1515-20.

21. Upadhyaya L, Singh J, Agarwal V, Tewari RP. The implications of recent advances in

carboxymethyl chitosan based targeted drug delivery and tissue engineering applications.

J Control Release. 2014;186:54-87.

22. Shrestha A, Shi Z, Neoh KG, Kishen A. Nanoparticulates for antibiofilm treatment

and effect of aging on its antibacterial activity. J Endod. 2010;36(6):1030-5.

23. Pulavendran S, Rose C, Mandal AB. Hepatocyte growth factor incorporated chitosan

nanoparticles augment the differentiation of stem cell into hepatocytes for the recovery of

liver cirrhosis in mice. J Nanobiotechnology. 2011;9:15.

Page 33: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

25  

24. Parajo Y, D'Angelo I, Welle A, Garcia-Fuentes M, Alonso MJ. Hyaluronic

acid/Chitosan nanoparticles as delivery vehicles for VEGF and PDGF-BB. Drug Deliv.

2010;17(8):596-604.

25. Shrestha S, Diogenes A, Kishen A. Temporal-controlled Release of Bovine Serum

Albumin from Chitosan Nanoparticles: Effect on the Regulation of Alkaline Phosphatase

Activity in Stem Cells from Apical Papilla. J Endod. 2014;40(9):1349-54.

26. Shrestha S, Diogenes A, Kishen A. Temporal-controlled Dexamethasone Releasing

Chitosan Nanoparticle System Enhances Odontogenic Differentiation of Stem Cells from

Apical Papilla. J Endod. 2015.

27. Zhu Y, Liu T, Song K, Jiang B, Ma X, Cui Z. Collagen-chitosan polymer as a

scaffold for the proliferation of human adipose tissue-derived stem cells. J Mater Sci

Mater Med. 2009;20(3):799-808.

28. Gorgieva S, Kokol V. Preparation, characterization, and in vitro enzymatic

degradation of chitosan-gelatine hydrogel scaffolds as potential biomaterials. J Biomed

Mater Res A. 2012;100(7):1655-67.

29. Ruparel NB, de Almeida JF, Henry MA, Diogenes A. Characterization of a stem cell

of apical papilla cell line: effect of passage on cellular phenotype. J Endod.

2013;39(3):357-63.

30. Lee JE, Kim KE, Kwon IC, Ahn HJ, Lee SH, Cho H, et al. Effects of the controlled-

released TGF-beta 1 from chitosan microspheres on chondrocytes cultured in a

collagen/chitosan/glycosaminoglycan scaffold. Biomaterials. 2004;25(18):4163-73.

31. Cohen S, Bano MC, Cima LG, Allcock HR, Vacanti JP, Vacanti CA, et al. Design of

synthetic polymeric structures for cell transplantation and tissue engineering. Clin Mater.

1993;13(1-4):3-10.

32. Tachibana A, Nishikawa Y, Nishino M, Kaneko S, Tanabe T, Yamauchi K. Modified

keratin sponge: binding of bone morphogenetic protein-2 and osteoblast differentiation. J

Biosci Bioeng. 2006;102(5):425-9.

33. Kim H, Kim HW, Suh H. Sustained release of ascorbate-2-phosphate and

dexamethasone from porous PLGA scaffolds for bone tissue engineering using

mesenchymal stem cells. Biomaterials. 2003;24(25):4671-9.

Page 34: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

26  

34. Sonoyama W, Liu Y, Fang D, Yamaza T, Seo BM, Zhang C, et al. Mesenchymal

stem cell-mediated functional tooth regeneration in swine. PLoS One. 2006;1:e79.

35. Diogenes A, Henry MA, Teixeira FB, Hargreaves KM. An update on clinical

regenerative endodontics. Endodontic Topics. 2013;28:2-23.

36. Smith AJ. Vitality of the dentin-pulp complex in health and disease: growth factors as

key mediators. J Dent Educ. 2003;67(6):678-89.

37. Tziafas D, Smith AJ, Lesot H. Designing new treatment strategies in vital pulp

therapy. J Dent. 2000;28(2):77-92.

38. Melin M, Joffre-Romeas A, Farges JC, Couble ML, Magloire H, Bleicher F. Effects

of TGFbeta1 on dental pulp cells in cultured human tooth slices. J Dent Res.

2000;79(9):1689-96.

39. Ochiai H, Yamamoto Y, Yokoyama A, Yamashita H, Matsuzaka K, Abe S, et al. Dual

Nature of TGF-β1 in Osteoblastic Differentiation of Human Periodontal Ligament Cells.

Journal of Hard Tissue Biology. 2010;19(3):187-94.

40. Wakefield LM, Winokur TS, Hollands RS, Christopherson K, Levinson AD, Sporn

MB. Recombinant latent transforming growth factor beta 1 has a longer plasma half-life

in rats than active transforming growth factor beta 1, and a different tissue distribution. J

Clin Invest. 1990;86(6):1976-84.

 

 

 

   

   

 

   

Page 35: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

27  

Chapter 4 Discussion, Conclusion and Future Directions

4

4.1 General Discussion A scaffold capable of attracting stem cells and delivering BM in a temporal-controlled

manner is essential for optimizing tissue engineering. In this study, we described a

CMCS scaffold containing controlled TGF-β1-releasing CSnp that may have practical

application to regenerative endodontics. The engineered matrix/nanoparticle scaffold

mimicked dental extracellular matrix, possessed high porosity, and significantly

enhanced SCAP migration and differentiation.

A properly engineered scaffold may serve as an initial attachment site for cells and BM

as well as structural support for immature tissue. Scaffolds in successful cell-free tissue

engineering applications should exhibit certain biophysical characteristics and deliver

BM to attract stem cells and promote their growth and differentiation. Scaffold porosity

facilitates diffusion of nutrients, clearance of degradation/cellular byproducts and cell

growth (45). The degree of crosslinking and porosity influences the rate of degradation of

a scaffold, which in turn, has been shown to affect the rate of release of immobilized

biomolecules over time (101, 143).

Scaffold degradation in vivo should be congruent with the new tissue growth, allowing

continued structural support and having little or no negative impact on the host tissue.

The impact of scaffold degradation byproducts on the residual functioning scaffold as

well as on the associated biological tissues deserves consideration. Undesirable evocation

of immune or inflammatory response, deterioration of scaffold characteristic properties

(e.g. stiffness, porosity, degradation rate), and host cell toxicity are legitimate concerns as

breakdown products of some materials have been shown to induce these negative effects

(49). Additionally, in the presence of infection, scaffold breakdown products should not

extend the inflammatory phase, which may delay healing.

The scaffolds used in this study consisted of chitosan and gelatin, two biopolymers

degraded by naturally occurring enzymes resident in human tissue, which show little to

Page 36: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

28  

no inflammatory response, and yield degradation products that include small sugar

molecules, peptides and amino acids well tolerated and processed by host cells (55, 137).

Moreover, chitosan demonstrates anti-bacterial and anti-inflammatory effects (74, 144).

The polycationic surface of chitosan allows adsorption of chitosan onto the anionic

bacterial cell surface, disrupting cell membrane integrity resulting in leakage of

intracellular components causing cell death. Chitosan also inhibits pro-inflammatory

cytokine production by host cells. It should be noted that changes in pH levels observed

during scaffold degradation in our study were negligible and remained at neutral levels.

Other scaffold materials have been shown to reduce the local tissue pH considerably (49).

Scaffolds fabricated by cross-linking gelatin with chitosan derivatives produced

improved mechanical properties and increased swelling capacity (137). Increases in

swelling capacity of the scaffold may be desirable for endodontic applications to improve

scaffold substantivity and adaptation to the root canal walls after implantation. Naturally,

concern must be directed to forces that may be exerted on the root wall and their potential

to initiate microcrack formation due to swelling of scaffold material placed inside the

root canal. The chitosan-gelatin scaffolds used in our study had dramatically low yield

strengths relative to dentin. Additionally, while swelling as a percentage of weight was

impressive in our study, the volumetric change was minimal and no significant increases

were detected at time points beyond 1 day. Furthermore, modifications to the scaffold

fabrication phase such as altering the gelatin content, scaffold porosity and degree of

crosslinking are examples of control over scaffold swelling and degradation to mitigate

this concern.

Favourable scaffold-cell interaction, as observed through SEM in our study, infers that

the CMCS scaffold used in this study was compatible with SCAP. SCAP are ecto-

mesenchymal stem cells from the apical papilla of developing human teeth and represent

a population of cells that can migrate to the root canal wall and participate in tissue

regeneration or organized repair. SCAP has demonstrated increased migration, tissue

regeneration capacity, and telomerase activity when compared to dental pulp stem cells

from the same tooth (120). It has also been suggested that SCAP may be better equipped

Page 37: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

29  

to survive and maintain their potential for differentiation in adverse, low oxygen

conditions (119).

TGF-β1 is a BM released from cells and incorporated into dentin and other non-

collagenous proteins in vivo. It has been identified as a promoter of odontogenic

differentiation (102, 103). However, availability of this molecule and other BM

sequestered in dentin matrix may not present in an optimum concentration or be available

for a duration necessary to satisfy stem cell requirements in tissue regeneration (139). In

the environment of a pulpless root canal, it is crucial that timely release of an optimum

concentration of growth factors be available for cells to survive, migrate and differentiate

(140). The importance of sustained release of BM for cell migration and differentiation

has previously been shown (113). Different concentrations of TGF-β1 were employed in

this study since it has been shown that TGF-β1 can effectively promote or inhibit cell

activity depending on the available concentration (100, 107). The significant increase in

SCAP viability compared to the same concentration of non-loaded CSnp or Free TGF-β1

was attributed to the release of bioactive TGF-β1 in a controlled manner over time. The

potential to provide such an environment was demonstrated in the TGF-β1-CSnp +

scaffold group where SCAP appeared to survive, migrate, and differentiate better than in

the other treatment groups.

TGF-β1 is known to modulate many cell activities including cell migration (140).

Significantly higher cell migration after 1 day in the Free TGF-β1 + scaffold group

compared with the TGF-β1-CSnp + scaffold group was expected since all of the TGF-β1

became available to the cells immediately when the TGF-β1 was in free form. After day

7, however, cell migration in the Free TGF-β1 + scaffold group decreased significantly,

which reflected a significant loss of bioactivity. In contrast, sustained release of TGF-β1

from TGF-β1-CSnp + scaffold group led to sustained cell migration over time. This was

an important finding in our study, as it showed the ability of the bioactive scaffold

containing TGF-β1-CSnp to address the issue of the short half-life of TGF-β1 (two to

three minutes) when it is used in free form in regenerative procedures (142).

Page 38: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

30  

Enhanced biomineralization of SCAP in the presence of TGF-β1-CSnp + scaffold was

also seen and can be attributed to the controlled release of TGF-β1 during the longer

periods of observation, as were the higher levels of DMP-1 and DSPP expression. DSPP

and DMP-1 are cytoplasmic proteins recognized as indicators of differentiation along

odontogenic lineages (145, 146). The immunofluorescent images also demonstrated more

uniform distribution of the cells in the TGF-β1-CSnp + scaffold group, when compared

to other experimental groups indicating that TGF-β1 released homogenously from the

CSnp dispersed in the scaffold.

In summary, biologically based regenerative endodontic procedures to address immature

teeth with pulp necrosis is a viable option. Increased success and outcome predictability

may be achievable by using a tissue engineering concept, which includes scaffolds, stem

cells, and bioactive molecules. Translatable findings using treatment strategies employing

materials that are biocompatible, anti-bacterial, anti-inflammatory and biodegradable may

offer another chairside option in the endodontic treatment of these teeth. Chitosan

appears to be an optimal material for this application due to its many positive attributes

previously described as well as its excellent functionality as a both a scaffold and

biomolecule delivery system as demonstrated in this study.

Page 39: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

31  

4.2 Conclusion In conclusion, the findings from the current study offered confirmation that CMCS

scaffold containing a sustained TGF-β1-releasing CSnp system:

• Possessed physicochemical characteristics conducive to SCAP survival and

attachment

• Promoted migration and enhanced differentiation of SCAP when compared to

Free TGF-β1 + scaffold, unloaded chitosan nanoparticles + scaffold, and scaffold

alone

• Illustrated the potential of chitosan-based scaffold to orchestrate BM delivery

with nanoparticle systems to facilitate dentin-pulp regeneration.

Page 40: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

32  

4.3 Future Directions Understanding both the role of individual BM and their reciprocal modulation in vivo will

be important to regenerative endodontics. Future research may include:

• Multiple BM release from polymeric scaffolds

• Modulation of scaffold/CSnp anti-bacterial/anti-inflammatory effect

• CSnp characterization over time in different tissue environments (e.g. pH,

temperature)

• Quality and spatial orientation of regenerated tissue

 

Page 41: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

33  

References 1. Holley RW. Control of growth of mammalian cells in cell culture. Nature.

1975;258(5535):487-90.

2. Roberts AB, Anzano MA, Wakefield LM, Roche NS, Stern DF, Sporn MB. Type beta

transforming growth factor: a bifunctional regulator of cellular growth. Proc Natl Acad

Sci U S A. 1985;82(1):119-23.

3. Chan BP, Leong KW. Scaffolding in tissue engineering: general approaches and

tissue-specific considerations. Eur Spine J. 2008;17(Suppl 4):467-79.

4. Fiske J, Davis DM, Leung KC, McMillan AS, Scott BJ. The emotional effects of

tooth loss in partially dentate people attending prosthodontic clinics in dental schools in

England, Scotland and Hong Kong: a preliminary investigation. Int Dent J.

2001;51(6):457-62.

5. Bergenholtz G. Effect of bacterial products on inflammatory reactions in the dental

pulp. Scand J Dent Res. 1977;85(2):122-9.

6. Lee BN, Moon JW, Chang HS, Hwang IN, Oh WM, Hwang YC. A review of the

regenerative endodontic treatment procedure. Restor Dent Endod. 2015;40(3):179-87.

7. Hargreaves KM, Diogenes A, Teixeira FB. Treatment options: biological basis of

regenerative endodontic procedures. J Endod. 2013;39(3 Suppl):S30-43.

8. Bose R, Nummikoski P, Hargreaves K. A retrospective evaluation of radiographic

outcomes in immature teeth with necrotic root canal systems treated with regenerative

endodontic procedures. J Endod. 2009;35(10):1343-9.

9. Ostby BN. The role of the blood clot in endodontic therapy. An experimental

histologic study. Acta Odontol Scand. 1961;19:324-53.

10. Nygaard-Ostby B, Hjortdal O. Tissue formation in the root canal following pulp

removal. Scand J Dent Res. 1971;79(5):333-49.

11. Vacanti JP, Morse MA, Saltzman WM, Domb AJ, Perez-Atayde A, Langer R.

Selective cell transplantation using bioabsorbable artificial polymers as matrices. J

Pediatr Surg. 1988;23(1 Pt 2):3-9.

12. Leach JB, Schmidt CE. Characterization of protein release from photocrosslinkable

hyaluronic acid-polyethylene glycol hydrogel tissue engineering scaffolds. Biomaterials.

2005;26(2):125-35.

Page 42: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

34  

13. Martin G, Ricucci D, Gibbs JL, Lin LM. Histological findings of

revascularized/revitalized immature permanent molar with apical periodontitis using

platelet-rich plasma. J Endod. 2013;39(1):138-44.

14. Lee DS, Yoon WJ, Cho ES, Kim HJ, Gronostajski RM, Cho MI, et al. Crosstalk

between nuclear factor I-C and transforming growth factor-β1 signaling regulates

odontoblast differentiation and homeostasis. PLoS One. 2011;6(12):e29160.

15. Kumar M. A review of chitin and chitosan applications. React Funct Polym.

2000;46(1):1-27.

16. Kumar MN, Muzzarelli RA, Muzzarelli C, Sashiwa H, Domb AJ. Chitosan chemistry

and pharmaceutical perspectives. Chem Rev. 2004;104(12):6017-84.

17. Sloan AJ, Smith AJ. Stem cells and the dental pulp: potential roles in dentine

regeneration and repair. Oral Dis. 2007;13(2):151-7.

18. AAE. Glossary of Endodontic Terms;2015. 8th Edition.

19. Shabahang S. Treatment options: apexogenesis and apexification. Pediatr Dent.

2013;35(2):125-8.

20. Hermann BW. Dentinobleration der Wurzelkanale nach der Behandlung mit Kalcium.

Zahnarzt Rundschau. 1930;39(888).

21. Mohammed M, Saujjanya KP, Jain D, Sangameshwar S. Role of Calcium Hydroxide

in Endodontics: A Review. Global Journal of Medicine and Public Health. 2012;1(1):66-

70.

22. Sjogren U, Figdor D, Spangberg L, Sundqvist G. The antimicrobial effect of calcium

hydroxide as a short-term intracanal dressing. Int Endod J. 1991;24(3):119-25.

23. Andreasen JO, Farik B, Munksgaard EC. Long-term calcium hydroxide as a root

canal dressing may increase risk of root fracture. Dent Traumatol. 2002;18(3):134-7.

24. Doyon GE, Dumsha T, von Fraunhofer JA. Fracture resistance of human root dentin

exposed to intracanal calcium hydroxide. J Endod. 2005;31(12):895-7.

25. Kawamoto R, Kurokawa H, Takubo C, Shimamura Y, Yoshida T, Miyazaki M.

Change in elastic modulus of bovine dentine with exposure to a calcium hydroxide paste.

J Dent. 2008;36(11):959-64.

Page 43: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

35  

26. Zarei M, Afkhami F, Malek Poor Z. Fracture resistance of human root dentin exposed

to calcium hydroxide intervisit medication at various time periods: an in vitro study. Dent

Traumatol. 2013;29(2):156-60.

27. Varghese L, Hegde M, Shetty A, Shetty C. Mineral Trioxide Aggregate: A Review.

Journal of Dental Sciences. 2014;2(2):19-22.

28. Torabinejad M, Chivian N. Clinical applications of mineral trioxide aggregate. J

Endod. 1999;25(3):197-205.

29. Parirokh M, Torabinejad M. Mineral trioxide aggregate: a comprehensive literature

review--Part III: Clinical applications, drawbacks, and mechanism of action. J Endod.

2010;36(3):400-13.

30. Jeeruphan T, Jantarat J, Yanpiset K, Suwannapan L, Khewsawai P, Hargreaves KM.

Mahidol study 1: comparison of radiographic and survival outcomes of immature teeth

treated with either regenerative endodontic or apexification methods: a retrospective

study. J Endod. 2012;38(10):1330-6.

31. Forsberg CM, Tedestam G. Traumatic injuries to teeth in Swedish children living in

an urban area. Swed Dent J. 1990;14(3):115-22.

32. Bryant SR. The effects of age, jaw site, and bone condition on oral implant outcomes.

The International Journal of Prosthodontics. 1997;11(5):470-90.

33. Mankani N, Chowdhary R, Patil BA, Nagaraj E, Madalli P. Osseointegrated Dental

Implants in Growing Children: A literature review. The Journal of Oral Implantology.

2014;40(5):627-31.

34. Robertson A, Andreasen FM, Bergenholtz G, Andreasen JO, Noren JG. Incidence of

pulp necrosis subsequent to pulp canal obliteration from trauma of permanent incisors. J

Endod. 1996;22(10):557-60.

35. Cvek M. Prognosis of luxated non-vital maxillary incisors treated with calcium

hydroxide and filled with gutta-percha. A retrospective clinical study. Endod Dent

Traumatol. 1992;8(2):45-55.

36. Diogenes A, Ruparel NB, Shiloah Y, Hargreaves KM. Regenerative endodontics: A

way forward. J Am Dent Assoc. 2016;147(5):372-80.

37. Tuan RS, Boland G, Tuli R. Adult mesenchymal stem cells and cell-based tissue

engineering. Arthritis Res Ther. 2003;5(1):32-45.

Page 44: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

36  

38. Muschler GF, Nakamoto C, Griffith LG. Engineering principles of clinical cell-based

tissue engineering. J Bone Joint Surg Am. 2004;86-a(7):1541-58.

39. Connolly J, Guse R, Lippiello L, Dehne R. Development of an osteogenic bone-

marrow preparation. J Bone Joint Surg Am. 1989;71(5):684-91.

40. Galler KM, Eidt A, Schmalz G. Cell-free approaches for dental pulp tissue

engineering. J Endod. 2014;40(4 Suppl):S41-5.

41. Mooney DJ, Powell C, Piana J, Rutherford B. Engineering dental pulp-like tissue in

vitro. Biotechnol Prog. 1996;12(6):865-8.

42. Iwaya SI, Ikawa M, Kubota M. Revascularization of an immature permanent tooth

with apical periodontitis and sinus tract. Dent Traumatol. 2001;17(4):185-7.

43. Banchs F, Trope M. Revascularization of immature permanent teeth with apical

periodontitis: new treatment protocol? J Endod. 2004;30(4):196-200.

44. Langer R, Folkman J. Polymers for the sustained release of proteins and other

macromolecules. Nature. 1976;263(5580):797-800.

45. Cohen S, Bano MC, Cima LG, Allcock HR, Vacanti JP, Vacanti CA, et al. Design of

synthetic polymeric structures for cell transplantation and tissue engineering. Clin Mater.

1993;13(1-4):3-10.

46. Wald HL, Sarakinos G, Lyman MD, Mikos AG, Vacanti JP, Langer R. Cell seeding

in porous transplantation devices. Biomaterials. 1993;14(4):270-8.

47. Mikos AG, Sarakinos G, Leite SM, Vacanti JP, Langer R. Laminated three-

dimensional biodegradable foams for use in tissue engineering. Biomaterials.

1993;14(5):323-30.

48. Taylor MS, Daniels AU, Andriano KP, Heller J. Six bioabsorbable polymers: in vitro

acute toxicity of accumulated degradation products. J Appl Biomater. 1994;5(2):151-7.

49. Athanasiou KA, Niederauer GG, Agrawal CM. Sterilization, toxicity,

biocompatibility and clinical applications of polylactic acid/polyglycolic acid

copolymers. Biomaterials. 1996;17(2):93-102.

50. Young CS, Terada S, Vacanti JP, Honda M, Bartlett JD, Yelick PC. Tissue

engineering of complex tooth structures on biodegradable polymer scaffolds. J Dent Res.

2002;81(10):695-700.

Page 45: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

37  

51. Zhang W, Walboomers XF, van Kuppevelt TH, Daamen WF, Bian Z, Jansen JA. The

performance of human dental pulp stem cells on different three-dimensional scaffold

materials. Biomaterials. 2006;27(33):5658-68.

52. Sumita Y, Honda MJ, Ohara T, Tsuchiya S, Sagara H, Kagami H, et al. Performance

of collagen sponge as a 3-D scaffold for tooth-tissue engineering. Biomaterials.

2006;27(17):3238-48.

53. Gotlieb EL, Murray PE, Namerow KN, Kuttler S, Garcia-Godoy F. An ultrastructural

investigation of tissue-engineered pulp constructs implanted within endodontically

treated teeth. J Am Dent Assoc. 2008;139(4):457-65.

54. Guo X, Park H, Young S, Kretlow JD, van den Beucken JJ, Baggett LS, et al. Repair

of osteochondral defects with biodegradable hydrogel composites encapsulating marrow

mesenchymal stem cells in a rabbit model. Acta Biomater. 2010;6(1):39-47.

55. Suh JK, Matthew HW. Application of chitosan-based polysaccharide biomaterials in

cartilage tissue engineering: a review. Biomaterials. 2000;21(24):2589-98.

56. Torabinejad M, Turman M. Revitalization of tooth with necrotic pulp and open apex

by using platelet-rich plasma: a case report. J Endod. 2011;37(2):265-8.

57. Bottino MC, Kamocki K, Yassen GH, Platt JA, Vail MM, Ehrlich Y, et al. Bioactive

nanofibrous scaffolds for regenerative endodontics. J Dent Res. 2013;92(11):963-9.

58. Petrino JA, Boda KK, Shambarger S, Bowles WR, McClanahan SB. Challenges in

regenerative endodontics: a case series. J Endod. 2010;36(3):536-41.

59. Rosa V, Zhang Z, Grande RH, Nor JE. Dental pulp tissue engineering in full-length

human root canals. J Dent Res. 2013;92(11):970-5.

60. Trevino EG, Patwardhan AN, Henry MA, Perry G, Dybdal-Hargreaves N, Hargreaves

KM, et al. Effect of irrigants on the survival of human stem cells of the apical papilla in a

platelet-rich plasma scaffold in human root tips. J Endod. 2011;37(8):1109-15.

61. Thomson A, Kahler B. Regenerative endodontics--biologically-based treatment for

immature permanent teeth: a case report and review of the literature. Aust Dent J.

2010;55(4):446-52.

62. Badylak SF. The extracellular matrix as a scaffold for tissue reconstruction. Semin

Cell Dev Biol. 2002;13(5):377-83.

Page 46: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

38  

63. Badylak SF, Freytes DO, Gilbert TW. Extracellular matrix as a biological scaffold

material: Structure and function. Acta Biomater. 2009;5(1):1-13.

64. Zhang Y, Zhang M. Three-dimensional macroporous calcium phosphate bioceramics

with nested chitosan sponges for load-bearing bone implants. J Biomed Mater Res.

2002;61(1):1-8.

65. Mastrogiacomo M, Scaglione S, Martinetti R, Dolcini L, Beltrame F, Cancedda R, et

al. Role of scaffold internal structure on in vivo bone formation in macroporous calcium

phosphate bioceramics. Biomaterials. 2006;27(17):3230-7.

66. Karageorgiou V, Kaplan D. Porosity of 3D biomaterial scaffolds and osteogenesis.

Biomaterials. 2005;26(27):5474-91.

67. Okuda K, Kawase T, Momose M, Murata M, Saito Y, Suzuki H, et al. Platelet-rich

plasma contains high levels of platelet-derived growth factor and transforming growth

factor-beta and modulates the proliferation of periodontally related cells in vitro. J

Periodontol. 2003;74(6):849-57.

68. Bernkop-Schnurch A. Chitosan and its derivatives: potential excipients for peroral

peptide delivery systems. Int J Pharm. 2000;194(1):1-13.

69. Galler KM, D'Souza RN, Hartgerink JD, Schmalz G. Scaffolds for dental pulp tissue

engineering. Adv Dent Res. 2011;23(3):333-9.

70. Shrestha A, Friedman S, Kishen A. Photodynamically crosslinked and chitosan-

incorporated dentin collagen. J Dent Res. 2011;90(11):1346-51.

71. Banerjee T, Mitra S, Kumar Singh A, Kumar Sharma R, Maitra A. Preparation,

characterization and biodistribution of ultrafine chitosan nanoparticles. Int J Pharm.

2002;243(1-2):93-105.

72. Agnihotri SA, Mallikarjuna NN, Aminabhavi TM. Recent advances on chitosan-

based micro- and nanoparticles in drug delivery. J Control Release. 2004;100(1):5-28.

73. Avadi M, Sadeghi M, Bayati K, Pouladzadeh M, Zohuriaan-Mehr M, al. e.

Diethylmethyl chitosan as an antibacterial agent: Synthesis, characterization and

antibacterial effects. Eur Polym J. 2004;40(7):1355-61.

74. Qi L, Xu Z, Jiang X, Hu C, Zou X. Preparation and antibacterial activity of chitosan

nanoparticles. Carbohydr Res. 2004;339(16):2693-700.

Page 47: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

39  

75. Sinha VR, Singla AK, Wadhawan S, Kaushik R, Kumria R, Bansal K, et al. Chitosan

microspheres as a potential carrier for drugs. Int J Pharm. 2004;274(1-2):1-33.

76. Rinaudo M. Chitin and chitosan: Properties and applications. Prog Polym Sci.

2006;31(7):603-32.

77. Shi Z, Neoh KG, Kang ET, Wang W. Antibacterial and mechanical properties of bone

cement impregnated with chitosan nanoparticles. Biomaterials. 2006;27(11):2440-9.

78. Chevrier A, Hoemann CD, Sun J, Buschmann MD. Chitosan-glycerol

phosphate/blood implants increase cell recruitment, transient vascularization and

subchondral bone remodeling in drilled cartilage defects. Osteoarthritis Cartilage.

2007;15(3):316-27.

79. Kim IY, Seo SJ, Moon HS, Yoo MK, Park IY, Kim BC, et al. Chitosan and its

derivatives for tissue engineering applications. Biotechnol Adv. 2008;26(1):1-21.

80. Shalumon K, Binulal N, Selvamurugan N, Nair S, Menon D, Furuike T, et al.

Electrospinning of carboxymethyl chitin/poly(vinyl alcohol) nanofibrous scaffolds for

tissue engineering applications. Carbohydr Polym. 2009;77(4):863-9.

81. Laudenslager MJ, Schiffman JD, Schauer CL. Carboxymethyl chitosan as a matrix

material for platinum, gold, and silver nanoparticles. Biomacromolecules.

2008;9(10):2682-5.

82. Tan W, Krishnaraj R, Desai TA. Evaluation of nanostructured composite collagen--

chitosan matrices for tissue engineering. Tissue Eng. 2001;7(2):203-10.

83. Kishen A, Shi Z, Shrestha A, Neoh KG. An investigation on the antibacterial and

antibiofilm efficacy of cationic nanoparticulates for root canal disinfection. J Endod.

2008;34(12):1515-20.

84. Upadhyaya L, Singh J, Agarwal V, Tewari RP. The implications of recent advances in

carboxymethyl chitosan based targeted drug delivery and tissue engineering applications.

J Control Release. 2014;186:54-87.

85. Shrestha A, Shi Z, Neoh KG, Kishen A. Nanoparticulates for antibiofilm treatment

and effect of aging on its antibacterial activity. J Endod. 2010;36(6):1030-5.

86. Schrieber R, Gareis H. Gelatine Handbook: Theory and Industrial Practice.

Weinheim: Weinheim:WILEY-VCH Verlag GmbH & Co. KGaA; 2007.

Page 48: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

40  

87. Dong Z, Wang Q, Du Y. Alginate/gelatin blend films and their properties for drug

controlled release. J Memb Sci. 2006;280(1-2):37-44.

88. Jatariu Cadinoiu AN, Popa M, Curteanu S, Peptu CA. Covalent and ionic co-cross-

linking--an original way to prepare chitosan-gelatin hydrogels for biomedical

applications. J Biomed Mater Res A. 2011;98(3):342-50.

89. Yorio T, Clark A, Wax M. Ocular Therapeutics: Eye on New Discoveries. First ed.

New York, NY, USA: Elsevier; 2008.

90. Eroglu H, Kas H, Oner L, Turkoglu O, Akalan N, Sargon M, et al. The in-vitro and

in-vivo characterization of PLGA:L-PLA microspheres containing dexamethasone

sodium phosphate. Journal of Microencapsulation. 2001;18(5):603-12.

91. Yoon JJ, Kim JH, Park TG. Dexamethasone-releasing biodegradable polymer

scaffolds fabricated by a gas-foaming/salt-leaching method. Biomaterials.

2003;24(13):2323-9.

92. Monteiro N, Martins A, Ribeiro D, Faria S, Fonseca N, Moreira J, et al. On the use of

dexamethasone-loaded liposomes to induce the osteogenic differentiation of human

mesenchymal stem cells. J Tissue Eng Regen Med 2013.

93. Nuttelman CR, Tripodi MC, Anseth KS. Dexamethasone-functionalized gels induce

osteogenic differentiation of encapsulated hMSCs. J Biomed Mater Res A.

2006;76(1):183-95.

94. Jaiswal N, Haynesworth SE, Caplan AI, Bruder SP. Osteogenic differentiation of

purified, culture-expanded human mesenchymal stem cells in vitro. J Cell Biochem.

1997;64(2):295-312.

95. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, Mosca JD, et al.

Multilineage potential of adult human mesenchymal stem cells. Science.

1999;284(5411):143-7.

96. Albuquerque MT, Valera MC, Nakashima M, Nor JE, Bottino MC. Tissue-

engineering-based strategies for regenerative endodontics. J Dent Res. 2014;93(12):1222-

31.

97. Kim JY, Xin X, Moioli EK, Chung J, Lee CH, Chen M, et al. Regeneration of dental-

pulp-like tissue by chemotaxis-induced cell homing. Tissue Eng Part A.

2010;16(10):3023-31.

Page 49: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

41  

98. Smith AJ, Duncan HF, Diogenes A, Simon S, Cooper PR. Exploiting the Bioactive

Properties of the Dentin-Pulp Complex in Regenerative Endodontics. J Endod.

2016;42(1):47-56.

99. Nakashima M, Reddi AH. The application of bone morphogenetic proteins to dental

tissue engineering. Nat Biotechnol. 2003;21(9):1025-32.

100. Bashutski JD, Wang HL. Role of platelet-rich plasma in soft tissue root-coverage

procedures: a review. Quintessence Int. 2008;39(6):473-83.

101. Tachibana A, Nishikawa Y, Nishino M, Kaneko S, Tanabe T, Yamauchi K.

Modified keratin sponge: binding of bone morphogenetic protein-2 and osteoblast

differentiation. J Biosci Bioeng. 2006;102(5):425-9.

102. Byun YK, Kim KH, Kim SH, Kim YS, Koo KT, Kim TI, et al. Effects of

immunosuppressants, FK506 and cyclosporin A, on the osteogenic differentiation of rat

mesenchymal stem cells. J Periodontal Implant Sci. 2012;42(3):73-80.

103. Smith AJ. Vitality of the dentin-pulp complex in health and disease: growth

factors as key mediators. J Dent Educ. 2003;67(6):678-89.

104. Chen YG. Endocytic regulation of TGF-beta signaling. Cell Res. 2009;19(1):58-

70.

105. Lu Z, Murray JT, Luo W, Li H, Wu X, Xu H, et al. Transforming growth factor

beta activates Smad2 in the absence of receptor endocytosis. J Biol Chem.

2002;277(33):29363-8.

106. Manning M, Patel K, Borchardt R. Stability of protein pharmaceuticals. Pharm

Res. 1989;6(11):903-18.

107. Lee JE, Kim KE, Kwon IC, Ahn HJ, Lee SH, Cho H, et al. Effects of the

controlled-released TGF-beta 1 from chitosan microspheres on chondrocytes cultured in a

collagen/chitosan/glycosaminoglycan scaffold. Biomaterials. 2004;25(18):4163-73.

108. Sassa Benedete AP, Sobral AP, Lima DM, Kamibeppu L, Soares FA, Lourenco

SV. Expression of transforming growth factor-beta 1, -beta 2, and -beta 3 in human

developing teeth: immunolocalization according to the odontogenesis phases. Pediatr Dev

Pathol. 2008;11(3):206-12.

Page 50: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

42  

109. Khil MS, Cha DI, Kim HY, Kim IS, Bhattarai N. Electrospun nanofibrous

polyurethane membrane as wound dressing. J Biomed Mater Res B Appl Biomater.

2003;67(2):675-9.

110. McNaught A, Wilkinson A. IUPAC. Compendium of Chemical Terminology.

Second ed. Oxford: Blackwell Scientific Publications; 2014.

111. Pulavendran S, Rose C, Mandal AB. Hepatocyte growth factor incorporated

chitosan nanoparticles augment the differentiation of stem cell into hepatocytes for the

recovery of liver cirrhosis in mice. J Nanobiotechnology. 2011;9:15.

112. Parajo Y, D'Angelo I, Welle A, Garcia-Fuentes M, Alonso MJ. Hyaluronic

acid/Chitosan nanoparticles as delivery vehicles for VEGF and PDGF-BB. Drug Deliv.

2010;17(8):596-604.

113. Shrestha S, Diogenes A, Kishen A. Temporal-controlled release of bovine serum

albumin from chitosan nanoparticles: effect on the regulation of alkaline phosphatase

activity in stem cells from apical papilla. J Endod. 2014;40(9):1349-54.

114. Shrestha S, Diogenes A, Kishen A. Temporal-controlled Dexamethasone

Releasing Chitosan Nanoparticle System Enhances Odontogenic Differentiation of Stem

Cells from Apical Papilla. J Endod. 2015.

115. Egusa H, Sonoyama W, Nishimura M, Atsuta I, Akiyama K. Stem cells in

dentistry--part I: stem cell sources. J Prosthodont Res. 2012;56(3):151-65.

116. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, Krause D, et

al. Minimal criteria for defining multipotent mesenchymal stromal cells. The

International Society for Cellular Therapy position statement. Cytotherapy.

2006;8(4):315-7.

117. Huang GT, Gronthos S, Shi S. Mesenchymal stem cells derived from dental

tissues vs. those from other sources: their biology and role in regenerative medicine. J

Dent Res. 2009;88(9):792-806.

118. Martin DE, De Almeida JF, Henry MA, Khaing ZZ, Schmidt CE, Teixeira FB, et

al. Concentration-dependent effect of sodium hypochlorite on stem cells of apical papilla

survival and differentiation. J Endod. 2014;40(1):51-5.

119. Diogenes A, Henry MA, Teixeira FB, Hargreaves KM. An update on clinical

regenerative endodontics. Endodontic Topics. 2013;28:2-23.

Page 51: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

43  

120. Sonoyama W, Liu Y, Fang D, Yamaza T, Seo BM, Zhang C, et al. Mesenchymal

stem cell-mediated functional tooth regeneration in swine. PLoS One. 2006;1:e79.

121. Xu L, Tang L, Jin F, Liu XH, Yu JH, Wu JJ, et al. The apical region of developing

tooth root constitutes a complex and maintains the ability to generate root and

periodontium-like tissues. J Periodontal Res. 2009;44(2):275-82.

122. Huang GT, Yamaza T, Shea LD, Djouad F, Kuhn NZ, Tuan RS, et al.

Stem/progenitor cell-mediated de novo regeneration of dental pulp with newly deposited

continuous layer of dentin in an in vivo model. Tissue Eng Part A. 2010;16(2):605-15.

123. Tziafas D, Kodonas K. Differentiation potential of dental papilla, dental pulp, and

apical papilla progenitor cells. J Endod. 2010;36(5):781-9.

124. Althumairy RI, Teixeira FB, Diogenes A. Effect of dentin conditioning with

intracanal medicaments on survival of stem cells of apical papilla. J Endod.

2014;40(4):521-5.

125. Ruparel NB, Teixeira FB, Ferraz CC, Diogenes A. Direct effect of intracanal

medicaments on survival of stem cells of the apical papilla. J Endod. 2012;38(10):1372-

5.

126. Karp JM, Leng Teo GS. Mesenchymal stem cell homing: the devil is in the

details. Cell Stem Cell. 2009;4(3):206-16.

127. Kim K, Lee CH, Kim BK, Mao JJ. Anatomically shaped tooth and periodontal

regeneration by cell homing. J Dent Res. 2010;89(8):842-7.

128. Yang J, Yuan G, Chen Z. Pulp Regeneration: Current Approaches and Future

Challenges. Front Physiol. 2016;7:58.

129. Suzuki T, Lee CH, Chen M, Zhao W, Fu SY, Qi JJ, et al. Induced migration of

dental pulp stem cells for in vivo pulp regeneration. J Dent Res. 2011;90(8):1013-8.

130. Yeom KH, Ariyoshi W, Okinaga T, Washio A, Morotomi T, Kitamura C, et al.

Platelet-rich plasma enhances the differentiation of dental pulp progenitor cells into

odontoblasts. Int Endod J. 2016;49(3):271-8.

131. Zhang DD, Chen X, Bao ZF, Chen M, Ding ZJ, Zhong M. Histologic comparison

between platelet-rich plasma and blood clot in regenerative endodontic treatment: an

animal study. J Endod. 2014;40(9):1388-93.

Page 52: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

44  

132. Jadhav GR, Shah N, Logani A. Platelet-rich plasma supplemented

revascularization of an immature tooth associated with a periapical lesion in a 40-year-

old man. Case Rep Dent. 2014;2014:479584.

133. Bezgin T, Yilmaz AD, Celik BN, Sonmez H. Concentrated platelet-rich plasma

used in root canal revascularization: 2 case reports. Int Endod J. 2014;47(1):41-9.

134. Choi BH, Zhu SJ, Kim BY, Huh JY, Lee SH, Jung JH. Effect of platelet-rich

plasma (PRP) concentration on the viability and proliferation of alveolar bone cells: an in

vitro study. Int J Oral Maxillofac Surg. 2005;34(4):420-4.

135. de Chevigny C, Dao TT, Basrani BR, Marquis V, Farzaneh M, Abitbol S, et al.

Treatment outcome in endodontics: the Toronto study--phase 4: initial treatment. J

Endod. 2008;34(3):258-63.

136. Zhu Y, Liu T, Song K, Jiang B, Ma X, Cui Z. Collagen-chitosan polymer as a

scaffold for the proliferation of human adipose tissue-derived stem cells. J Mater Sci

Mater Med. 2009;20(3):799-808.

137. Gorgieva S, Kokol V. Preparation, characterization, and in vitro enzymatic

degradation of chitosan-gelatine hydrogel scaffolds as potential biomaterials. J Biomed

Mater Res A. 2012;100(7):1655-67.

138. Ruparel NB, de Almeida JF, Henry MA, Diogenes A. Characterization of a stem

cell of apical papilla cell line: effect of passage on cellular phenotype. J Endod.

2013;39(3):357-63.

139. Tziafas D, Smith AJ, Lesot H. Designing new treatment strategies in vital pulp

therapy. J Dent. 2000;28(2):77-92.

140. Melin M, Joffre-Romeas A, Farges JC, Couble ML, Magloire H, Bleicher F.

Effects of TGFbeta1 on dental pulp cells in cultured human tooth slices. J Dent Res.

2000;79(9):1689-96.

141. Ochiai H, Yamamoto Y, Yokoyama A, Yamashita H, Matsuzaka K, Abe S, et al.

Dual Nature of TGF-β1 in Osteoblastic Differentiation of Human Periodontal Ligament

Cells. Journal of Hard Tissue Biology. 2010;19(3):187-94.

142. Wakefield LM, Winokur TS, Hollands RS, Christopherson K, Levinson AD,

Sporn MB. Recombinant latent transforming growth factor beta 1 has a longer plasma

Page 53: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

45  

half-life in rats than active transforming growth factor beta 1, and a different tissue

distribution. J Clin Invest. 1990;86(6):1976-84.

143. Kim H, Kim HW, Suh H. Sustained release of ascorbate-2-phosphate and

dexamethasone from porous PLGA scaffolds for bone tissue engineering using

mesenchymal stem cells. Biomaterials. 2003;24(25):4671-9.

144. Friedman AJ, Phan J, Schairer DO, Champer J, Qin M, Pirouz A, et al.

Antimicrobial and anti-inflammatory activity of chitosan-alginate nanoparticles: a

targeted therapy for cutaneous pathogens. J Invest Dermatol. 2013;133(5):1231-9.

145. MacDougall M, Simmons D, Luan X, Nydegger J, Feng J, Gu TT. Dentin

phosphoprotein and dentin sialoprotein are cleavage products expressed from a single

transcript coded by a gene on human chromosome 4. Dentin phosphoprotein DNA

sequence determination. J Biol Chem. 1997;272(2):835-42.

146. Wu G, Deng ZH, Fan XJ, Ma ZF, Sun YJ, Ma DD, et al. Odontogenic potential of

mesenchymal cells from hair follicle dermal papilla. Stem Cells Dev. 2009;18(4):583-9.

 

Page 54: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

46  

Appendix I  

       

 

0

200

400

600

800

1000

0 0.5 1 2 4 24

Swel

ling

ratio

(%)

Time (hours)

B A

0

10

20

30

40

50

60

0 7 14 21

Deg

rada

tion

(%)

Time (days)

C

D E

Figure 1. Characterization of scaffold: (A) Scaffold morphology as observed under SEM illustrating porous structure; (B) Scaffold swelling ratio; (C) Scaffold degradation (%) in the presence of lysozyme; (D) FTIR Spectra of CMCS, Gelatin, and the CMCS/gelatin scaffold; (E) SCAP morphology as observed under SEM illustrating favorable response to scaffold.

Page 55: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

47  

Figure 2: Characterization of TGF-β1-CSnp. (A) Cumulative release (ng) of TGF-β1 from TGF-β1-CSnp; (B) Relative viability of SCAP exposed to Free TGF-β1, TGF-β1-CSnp, or CSnp over 1, 3 and 14 days. (C) Representative fluorescent microscopic view after the calcein-AM staining (upper row, magnification 20X) and phase contrast microscopic images (lower row, magnification 40X). Data represent means ± SD (n = 3). *Significant difference between groups, p < 0.001.

0  

20  

40  

60  

80  

100  

120  

140  

1   3   14  

Relative  Viability  (%

 control)  

Time  (days)  

B

Free  TGF-­‐b1  

TGF-­‐b1-­‐CSnp  

CSnp  

Free TGF-β1

TGF-β1-CSnp CSnp

                   *                              *  

0 2 4 6 8

10 12 14 16

0 100 200 300 400 500 600 700

Cum

ulat

ive

rele

ase

(ng)

Time (hours)

A

C  

Page 56: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

48  

 

Figure 3: (A) TGF-β1-induced migration of SCAP: Cell migration was examined by a Transwell assay. SCAP were cultured in presence of Free TGF-β1, TGF-β1 released from TGF-β1-CSnp incubated for different time periods and CSnp. The lower membrane surfaces were photographed through a microscope at 20X magnification. Migrated cells in each field were counted. *Significant difference between groups, p < 0.028. (B) Effect of nanoparticles on biomineralization: ARS was quantified for biomineralization in SCAP culture for 3 weeks in presence of Free TGF-β1, TGF-β1-CSnp or CSnp. Data are shown as mean OD/µg of total protein. Data represent means ± SD (n = 3). *Significant difference between groups, p < 0.001.

0

0.001

0.002

0.003

0.004

0.005

0.006

1 week 2 week 3 week

OD

560/µ

g of

tot

al p

rote

in

Free TGF-b1

TGF-b1-CSnp

CSnp

B

7 14 21 Time (days)

Free TGF-β1 + scaffold TGF-β1-CSnp + scaffold

CSnp + scaffold

 *  

0

50

100

150

200

250

1 7 14 21

Mig

ratio

n (%

of c

ontr

ol)

Time (days)

Free TGF-b1

TGF-b1-CSnp

CSnp

A

Free TGF-β1 + scaffold TGF-β1-CSnp + scaffold

CSnp + scaffold

                       *  

               *    

                     *  

Page 57: Effects of a Bioactive Scaffold Containing a …...ii! Effects of a Bioactive Scaffold Containing a Sustained TGF-β1-Releasing Nanoparticle System on the Migration and Differentiation

 

 

49  

 

   

0  

5000  

10000  

15000  

20000  

25000  

Free  TGF-­‐β1  +  scaffold   TGF-­‐β1-­‐CSnp  +  scaffold   CSnp  +  scaffold   Scaffold  

Relative  Fluorescence  

 (arbitrary  units)  

DMP-­‐1  

DSPP  

B  *          *  

DMP-1 DSPP

A  

Figure 4: Immunofluorescence of DMP-1 and DSPP during odontoblast differentiation: (A) FITC-conjugated antibody was used to detect the localization of the protein (green signal); all samples were counterstained with DAPI (blue signal). (B) Quantification of protein expression by ImageJ software and data are presented as relative mean fluorescence intensity. *Significant difference between groups, p < 0.001.